• Title/Summary/Keyword: Chloride intracellular channel (CLIC1)

Search Result 2, Processing Time 0.015 seconds

The Binding of Human CLIC1 with SEDL and Its Characterization in vitro

  • Park, Jeong-Soon;Lee, Kyoung-Mi;Jeong, Mi-Suk;Jin, Gyoung-Ean;Jang, Se-Bok
    • Bulletin of the Korean Chemical Society
    • /
    • v.28 no.4
    • /
    • pp.574-580
    • /
    • 2007
  • Full-length chloride intracellular channel protein 1 (CLIC1) is a member of the family of proteins related to bovine chloride intracellular channel p64. Mutations in the SEDL gene cause spondyloepiphyseal dysplasia tarda (SEDT), a rare X-linked chondrodysplasia. The link between the intracellular chloride channels and SEDL is an important step toward understanding their functional interplay. In the present study, CLIC1 protein was subcloned into the pGEX-KG vector and overexpressed in XL-1 blue cells. We developed a large-scale expression system composed of glutathione S-transferase (GST) fused with a 240-amino-acid CLIC1 protein in Escherichia coli. The soluble CLIC1 protein was successfully purified to homogeneity, and its purity, identity, activity and conformation were determined using SDS-PAGE, MALDI-MS, biophotometer and circular dichroism spectroscopic studies. The binding of both CLIC1 and SEDL proteins in vitro was detected by BIAcore biosensor and fluorescence measurements.

Extracellular Acidification Augments NLRP3-Mediated Inflammasome Signaling in Macrophages

  • Byeong Jun Chae;Kyung-Seo Lee;Inhwa Hwang;Je-Wook Yu
    • IMMUNE NETWORK
    • /
    • v.23 no.3
    • /
    • pp.23.1-23.17
    • /
    • 2023
  • Inflammation is a series of host defense processes in response to microbial infection and tissue injury. Inflammatory processes frequently cause extracellular acidification in the inflamed region through increased glycolysis and lactate secretion. Therefore, the immune cells infiltrating the inflamed region encounter an acidic microenvironment. Extracellular acidosis can modulate the innate immune response of macrophages; however, its role for inflammasome signaling still remains elusive. In the present study, we demonstrated that macrophages exposed to an acidic microenvironment exhibited enhanced caspase-1 processing and IL-1β secretion compared with those under physiological pH. Moreover, exposure to an acidic pH increased the ability of macrophages to assemble the NLR family pyrin domain containing 3 (NLRP3) inflammasome in response to an NLRP3 agonist. This acidosis-mediated augmentation of NLRP3 inflammasome activation occurred in bone marrow-derived macrophages but not in bone marrow-derived neutrophils. Notably, exposure to an acidic environment caused a reduction in the intracellular pH of macrophages but not neutrophils. Concordantly, macrophages, but not neutrophils, exhibited NLRP3 agonist-mediated translocation of chloride intracellular channel protein 1 (CLIC1) into their plasma membranes under an acidic microenvironment. Collectively, our results demonstrate that extracellular acidosis during inflammation can increase the sensitivity of NLRP3 inflammasome formation and activation in a CLIC1-dependent manner. Thus, CLIC1 may be a potential therapeutic target for NLRP3 inflammasome-mediated pathological conditions.